451
|
Spella M, Ntaliarda G, Skiadas G, Lamort AS, Vreka M, Marazioti A, Lilis I, Bouloukou E, Giotopoulou GA, Pepe MAA, Weiss SAI, Petrera A, Hauck SM, Koch I, Lindner M, Hatz RA, Behr J, Arendt KAM, Giopanou I, Brunn D, Savai R, Jenne DE, de Château M, Yull FE, Blackwell TS, Stathopoulos GT. Non-Oncogene Addiction of KRAS-Mutant Cancers to IL-1β via Versican and Mononuclear IKKβ. Cancers (Basel) 2023; 15:1866. [PMID: 36980752 PMCID: PMC10047096 DOI: 10.3390/cancers15061866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Kirsten rat sarcoma virus (KRAS)-mutant cancers are frequent, metastatic, lethal, and largely undruggable. While interleukin (IL)-1β and nuclear factor (NF)-κB inhibition hold promise against cancer, untargeted treatments are not effective. Here, we show that human KRAS-mutant cancers are addicted to IL-1β via inflammatory versican signaling to macrophage inhibitor of NF-κB kinase (IKK) β. Human pan-cancer and experimental NF-κB reporter, transcriptome, and proteome screens reveal that KRAS-mutant tumors trigger macrophage IKKβ activation and IL-1β release via secretory versican. Tumor-specific versican silencing and macrophage-restricted IKKβ deletion prevents myeloid NF-κB activation and metastasis. Versican and IKKβ are mutually addicted and/or overexpressed in human cancers and possess diagnostic and prognostic power. Non-oncogene KRAS/IL-1β addiction is abolished by IL-1β and TLR1/2 inhibition, indicating cardinal and actionable roles for versican and IKKβ in metastasis.
Collapse
Affiliation(s)
- Magda Spella
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Giannoula Ntaliarda
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgios Skiadas
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Anne-Sophie Lamort
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Malamati Vreka
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Antonia Marazioti
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioannis Lilis
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Eleni Bouloukou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgia A. Giotopoulou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Mario A. A. Pepe
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Stefanie A. I. Weiss
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Agnese Petrera
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Ina Koch
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Michael Lindner
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Rudolph A. Hatz
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Juergen Behr
- Department of Internal Medicine V, Ludwig-Maximilian-University of Munich, 81377 Munich, Germany
| | - Kristina A. M. Arendt
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioanna Giopanou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
- Department of Internal Medicine and Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Dieter E. Jenne
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Max-Planck-Institute of Neurobiology, 82152 Planegg, Germany
| | | | - Fiona E. Yull
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Timothy S. Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Georgios T. Stathopoulos
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
452
|
Portal C, Lin Y, Rastogi V, Peterson C, Yiu SCH, Foster JW, Wilkerson A, Butovich IA, Iomini C. Primary cilia control cellular patterning of Meibomian glands during morphogenesis but not lipid composition. Commun Biol 2023; 6:282. [PMID: 36932132 PMCID: PMC10023665 DOI: 10.1038/s42003-023-04632-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
Meibomian glands (MGs) are modified sebaceous glands producing the tear film's lipids. Despite their critical role in maintaining clear vision, the mechanisms underlying MG morphogenesis in development and disease remain obscure. Cilia-mediate signals are critical for the development of skin adnexa, including sebaceous glands. Thus, we investigated the role of cilia in MG morphogenesis during development. Most cells were ciliated during early MG development, followed by cilia disassembly during differentiation. In mature glands, ciliated cells were primarily restricted to the basal layer of the proximal gland central duct. Cilia ablation in keratine14-expressing tissue disrupted the accumulation of proliferative cells at the distal tip but did not affect the overall rate of proliferation or apoptosis. Moreover, impaired cellular patterning during elongation resulted in hypertrophy of mature MGs with increased meibum volume without altering its lipid composition. Thus, cilia signaling networks provide a new platform to design therapeutic treatments for MG dysfunction.
Collapse
Affiliation(s)
- Céline Portal
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Yvonne Lin
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Varuni Rastogi
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Cornelia Peterson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Samuel Chi-Hung Yiu
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - James W Foster
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Amber Wilkerson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Igor A Butovich
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Carlo Iomini
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
| |
Collapse
|
453
|
Bailey LRJ, Bugg D, Reichardt IM, Ortaç CD, Gunaje J, Johnson R, MacCoss MJ, Sakamoto T, Kelly DP, Regnier M, Davis JM. MBNL1 regulates programmed postnatal switching between regenerative and differentiated cardiac states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532974. [PMID: 36993225 PMCID: PMC10055038 DOI: 10.1101/2023.03.16.532974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Discovering determinants of cardiomyocyte maturity and the maintenance of differentiated states is critical to both understanding development and potentially reawakening endogenous regenerative programs in adult mammalian hearts as a therapeutic strategy. Here, the RNA binding protein Muscleblind-like 1 (MBNL1) was identified as a critical regulator of cardiomyocyte differentiated states and their regenerative potential through transcriptome-wide control of RNA stability. Targeted MBNL1 overexpression early in development prematurely transitioned cardiomyocytes to hypertrophic growth, hypoplasia, and dysfunction, whereas loss of MBNL1 function increased cardiomyocyte cell cycle entry and proliferation through altered cell cycle inhibitor transcript stability. Moreover, MBNL1-dependent stabilization of the estrogen-related receptor signaling axis was essential for maintaining cardiomyocyte maturity. In accordance with these data, modulating MBNL1 dose tuned the temporal window of cardiac regeneration, where enhanced MBNL1 activity arrested myocyte proliferation, and MBNL1 deletion promoted regenerative states with prolonged myocyte proliferation. Collectively these data suggest MBNL1 acts as a transcriptome-wide switch between regenerative and mature myocyte states postnatally and throughout adulthood.
Collapse
|
454
|
Ray AT, Soriano P. FGF signaling regulates salivary gland branching morphogenesis by modulating cell adhesion. Development 2023; 150:dev201293. [PMID: 36861436 PMCID: PMC10112918 DOI: 10.1242/dev.201293] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Loss of FGF signaling leads to defects in salivary gland branching, but the mechanisms underlying this phenotype remain largely unknown. We disrupted expression of Fgfr1 and Fgfr2 in salivary gland epithelial cells and found that both receptors function coordinately in regulating branching. Strikingly, branching morphogenesis in double knockouts is restored by Fgfr1 and Fgfr2 (Fgfr1/2) knock-in alleles incapable of engaging canonical RTK signaling, suggesting that additional FGF-dependent mechanisms play a role in salivary gland branching. Fgfr1/2 conditional null mutants showed defective cell-cell and cell-matrix adhesion, both of which have been shown to play instructive roles in salivary gland branching. Loss of FGF signaling led to disordered cell-basement membrane interactions in vivo as well as in organ culture. This was partially restored upon introducing Fgfr1/2 wild-type or signaling alleles that are incapable of eliciting canonical intracellular signaling. Together, our results identify non-canonical FGF signaling mechanisms that regulate branching morphogenesis through cell-adhesion processes.
Collapse
Affiliation(s)
- Ayan T. Ray
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
455
|
Nunley H, Shao B, Grover P, Singh J, Joyce B, Kim-Yip R, Kohrman A, Watters A, Gal Z, Kickuth A, Chalifoux M, Shvartsman S, Posfai E, Brown LM. A novel ground truth dataset enables robust 3D nuclear instance segmentation in early mouse embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532646. [PMID: 36993260 PMCID: PMC10055179 DOI: 10.1101/2023.03.14.532646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
For investigations into fate specification and cell rearrangements in live images of preimplantation embryos, automated and accurate 3D instance segmentation of nuclei is invaluable; however, the performance of segmentation methods is limited by the images' low signal-to-noise ratio and high voxel anisotropy and the nuclei's dense packing and variable shapes. Supervised machine learning approaches have the potential to radically improve segmentation accuracy but are hampered by a lack of fully annotated 3D data. In this work, we first establish a novel mouse line expressing near-infrared nuclear reporter H2B-miRFP720. H2B-miRFP720 is the longest wavelength nuclear reporter in mice and can be imaged simultaneously with other reporters with minimal overlap. We then generate a dataset, which we call BlastoSPIM, of 3D microscopy images of H2B-miRFP720-expressing embryos with ground truth for nuclear instance segmentation. Using BlastoSPIM, we benchmark the performance of five convolutional neural networks and identify Stardist-3D as the most accurate instance segmentation method across preimplantation development. Stardist-3D, trained on BlastoSPIM, performs robustly up to the end of preimplantation development (> 100 nuclei) and enables studies of fate patterning in the late blastocyst. We, then, demonstrate BlastoSPIM's usefulness as pre-train data for related problems. BlastoSPIM and its corresponding Stardist-3D models are available at: blastospim.flatironinstitute.org.
Collapse
Affiliation(s)
- Hayden Nunley
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Binglun Shao
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
| | - Prateek Grover
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Jaspreet Singh
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Rebecca Kim-Yip
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Abraham Kohrman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Aaron Watters
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Zsombor Gal
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Alison Kickuth
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Madeleine Chalifoux
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Stanislav Shvartsman
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Lisa M. Brown
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| |
Collapse
|
456
|
Vanderkruk B, Maeshima N, Pasula DJ, An M, McDonald CL, Suresh P, Luciani DS, Lynn FC, Hoffman BG. Methylation of histone H3 lysine 4 is required for maintenance of beta cell function in adult mice. Diabetologia 2023; 66:1097-1115. [PMID: 36912927 PMCID: PMC10163146 DOI: 10.1007/s00125-023-05896-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023]
Abstract
AIMS/HYPOTHESIS Beta cells control glucose homeostasis via regulated production and secretion of insulin. This function arises from a highly specialised gene expression programme that is established during development and then sustained, with limited flexibility, in terminally differentiated cells. Dysregulation of this programme is seen in type 2 diabetes but mechanisms that preserve gene expression or underlie its dysregulation in mature cells are not well resolved. This study investigated whether methylation of histone H3 lysine 4 (H3K4), a marker of gene promoters with unresolved functional importance, is necessary for the maintenance of mature beta cell function. METHODS Beta cell function, gene expression and chromatin modifications were analysed in conditional Dpy30 knockout mice, in which H3K4 methyltransferase activity is impaired, and in a mouse model of diabetes. RESULTS H3K4 methylation maintains expression of genes that are important for insulin biosynthesis and glucose responsiveness. Deficient methylation of H3K4 leads to a less active and more repressed epigenome profile that locally correlates with gene expression deficits but does not globally reduce gene expression. Instead, developmentally regulated genes and genes in weakly active or suppressed states particularly rely on H3K4 methylation. We further show that H3K4 trimethylation (H3K4me3) is reorganised in islets from the Leprdb/db mouse model of diabetes in favour of weakly active and disallowed genes at the expense of terminal beta cell markers with broad H3K4me3 peaks. CONCLUSIONS/INTERPRETATION Sustained methylation of H3K4 is critical for the maintenance of beta cell function. Redistribution of H3K4me3 is linked to gene expression changes that are implicated in diabetes pathology.
Collapse
Affiliation(s)
- Ben Vanderkruk
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Nina Maeshima
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J Pasula
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Meilin An
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Cassandra L McDonald
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Priya Suresh
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Dan S Luciani
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Francis C Lynn
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Brad G Hoffman
- Diabetes Research Group, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
457
|
Lin Y, Banno K, Gil CH, Myslinski J, Hato T, Shelley WC, Gao H, Xuei X, Liu Y, Basile DP, Yoshimoto M, Prasain N, Tarnawsky SP, Adams RH, Naruse K, Yoshida J, Murphy MP, Horie K, Yoder MC. Origin, prospective identification, and function of circulating endothelial colony-forming cells in mice and humans. JCI Insight 2023; 8:e164781. [PMID: 36692963 PMCID: PMC10077473 DOI: 10.1172/jci.insight.164781] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Most circulating endothelial cells are apoptotic, but rare circulating endothelial colony-forming cells (C-ECFCs), also known as blood outgrowth endothelial cells, with proliferative and vasculogenic activity can be cultured; however, the origin and naive function of these C-ECFCs remains obscure. Herein, detailed lineage tracing revealed murine C-ECFCs emerged in the early postnatal period, displayed high vasculogenic potential with enriched frequency of clonal proliferative cells compared with tissue-resident ECFCs, and were not committed to or derived from the BM hematopoietic system but from tissue-resident ECFCs. In humans, C-ECFCs were present in the CD34bright cord blood mononuclear subset, possessed proliferative potential and in vivo vasculogenic function in a naive or cultured state, and displayed a single cell transcriptome sharing some umbilical venous endothelial cell features, such as a higher protein C receptor and extracellular matrix gene expression. This study provides an advance for the field by identifying the origin, naive function, and antigens to prospectively isolate C-ECFCs for translational studies.
Collapse
Affiliation(s)
- Yang Lin
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kimihiko Banno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | - Chang-Hyun Gil
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery
| | | | | | - William C. Shelley
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, and
| | | | - Yunlong Liu
- Department of Medical and Molecular Genetics, and
| | - David P. Basile
- Department of Anatomy Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Momoko Yoshimoto
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nutan Prasain
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stefan P. Tarnawsky
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ralf H. Adams
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Katsuhiko Naruse
- Department of Obstetrics & Gynecology, Nara Medical University, Kashihara, Nara, Japan
| | - Junko Yoshida
- Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | | | - Kyoji Horie
- Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
458
|
Yang J, Bergdorf K, Yan C, Luo W, Chen SC, Ayers D, Liu Q, Liu X, Boothby M, Groves SM, Oleskie AN, Zhang X, Maeda DY, Zebala JA, Quaranta V, Richmond A. CXCR2 expression during melanoma tumorigenesis controls transcriptional programs that facilitate tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.22.529548. [PMID: 36865260 PMCID: PMC9980137 DOI: 10.1101/2023.02.22.529548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Background Though the CXCR2 chemokine receptor is known to play a key role in cancer growth and response to therapy, a direct link between expression of CXCR2 in tumor progenitor cells during induction of tumorigenesis has not been established. Methods To characterize the role of CXCR2 during melanoma tumorigenesis, we generated tamoxifen-inducible tyrosinase-promoter driven Braf V600E /Pten -/- /Cxcr2 -/- and NRas Q61R /INK4a -/- /Cxcr2 -/- melanoma models. In addition, the effects of a CXCR1/CXCR2 antagonist, SX-682, on melanoma tumorigenesis were evaluated in Braf V600E /Pten -/- and NRas Q61R /INK4a -/- mice and in melanoma cell lines. Potential mechanisms by which Cxcr2 affects melanoma tumorigenesis in these murine models were explored using RNAseq, mMCP-counter, ChIPseq, and qRT-PCR; flow cytometry, and reverse phosphoprotein analysis (RPPA). Results Genetic loss of Cxcr2 or pharmacological inhibition of CXCR1/CXCR2 during melanoma tumor induction resulted in key changes in gene expression that reduced tumor incidence/growth and increased anti-tumor immunity. Interestingly, after Cxcr2 ablation, Tfcp2l1 , a key tumor suppressive transcription factor, was the only gene significantly induced with a log 2 fold-change greater than 2 in these three different melanoma models. Conclusions Here, we provide novel mechanistic insight revealing how loss of Cxcr2 expression/activity in melanoma tumor progenitor cells results in reduced tumor burden and creation of an anti-tumor immune microenvironment. This mechanism entails an increase in expression of the tumor suppressive transcription factor, Tfcp2l1, along with alteration in the expression of genes involved in growth regulation, tumor suppression, stemness, differentiation, and immune modulation. These gene expression changes are coincident with reduction in the activation of key growth regulatory pathways, including AKT and mTOR.
Collapse
|
459
|
Lav R, Krivanek J, Anthwal N, Tucker AS. Wnt signaling from Gli1-expressing apical stem/progenitor cells is essential for the coordination of tooth root development. Stem Cell Reports 2023; 18:1015-1029. [PMID: 36931279 PMCID: PMC10147554 DOI: 10.1016/j.stemcr.2023.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
Stem cell regulation plays a crucial role during development and homeostasis. Here, an essential source of Wnts from Gli1+ stem/progenitor cells was identified in the murine molar. Loss of Wnt production in Gli1+ apical stem/progenitor cells led to loss of Axin2 at the root apex, mis-regulation of SOX9, loss of BMP and Hh signaling, and truncation of root development. In the absence of Wnt signals, the root epithelium lost its integrity and epithelial identity. This phenotype could be partially mimicked by loss of Sox9 in the Gli1 population. Stabilization of Wnt signaling in the apical papilla led to rapid unordered differentiation of hard tissues and fragmentation of the epithelial root sheath. Wnt signaling from Gli1+ stem/progenitor cells, therefore, orchestrates root development, coordinating mesenchymal and epithelial interactions via SOX9 to regulate stem/progenitor cell expansion and differentiation. Our results demonstrate that disparate stem/progenitor cell populations are unified in their fundamental signaling interactions.
Collapse
Affiliation(s)
- Rupali Lav
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Neal Anthwal
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
| |
Collapse
|
460
|
Gu D, Soepriatna AH, Zhang W, Li J, Zhao J, Zhang X, Shu X, Wang Y, Landis BJ, Goergen CJ, Xie J. Activation of the Hedgehog signaling pathway leads to fibrosis in aortic valves. Cell Biosci 2023; 13:43. [PMID: 36864465 PMCID: PMC9983197 DOI: 10.1186/s13578-023-00980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/03/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Fibrosis is a pathological wound healing process characterized by excessive extracellular matrix deposition, which interferes with normal organ function and contributes to ~ 45% of human mortality. Fibrosis develops in response to chronic injury in nearly all organs, but the a cascade of events leading to fibrosis remains unclear. While hedgehog (Hh) signaling activation has been associated with fibrosis in the lung, kidney, and skin, it is unknown whether hedgehog signaling activation is the cause or the consequence of fibrosis. We hypothesize that activation of hedgehog signaling is sufficient to drive fibrosis in mouse models. RESULTS In this study, we provide direct evidence to show that activation of Hh signaling via expression of activated smoothened, SmoM2, is sufficient to induce fibrosis in the vasculature and aortic valves. We showed that activated SmoM2 -induced fibrosis is associated with abnormal function of aortic valves and heart. The relevance of this mouse model to human health is reflected in our findings that elevated GLI expression is detected in 6 out of 11 aortic valves from patients with fibrotic aortic valves. CONCLUSIONS Our data show that activating hedgehog signaling is sufficient to drive fibrosis in mice, and this mouse model is relevant to human aortic valve stenosis.
Collapse
Affiliation(s)
- Dongsheng Gu
- grid.257413.60000 0001 2287 3919Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, 1040 W. Walnut Street., Indianapolis, IN 46202 USA
| | - Arvin H. Soepriatna
- grid.169077.e0000 0004 1937 2197Purdue University Weldon School of Biomedical Engineering, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907 USA ,grid.40263.330000 0004 1936 9094School of Engineering, Center for Biomedical Engineering, Brown University, 184 Hope Street, Providence, RI 02912 USA
| | - Wenjun Zhang
- grid.257413.60000 0001 2287 3919Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, 1040 W. Walnut Street., Indianapolis, IN 46202 USA
| | - Jun Li
- grid.413087.90000 0004 1755 3939Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Jenny Zhao
- grid.257413.60000 0001 2287 3919Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, 1040 W. Walnut Street., Indianapolis, IN 46202 USA ,grid.189504.10000 0004 1936 7558Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118 USA
| | - Xiaoli Zhang
- grid.257413.60000 0001 2287 3919Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, 1040 W. Walnut Street., Indianapolis, IN 46202 USA
| | - Xianhong Shu
- grid.413087.90000 0004 1755 3939Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Yongshi Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Benjamin J. Landis
- grid.257413.60000 0001 2287 3919Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, 1040 W. Walnut Street., Indianapolis, IN 46202 USA
| | - Craig J. Goergen
- grid.169077.e0000 0004 1937 2197Purdue University Weldon School of Biomedical Engineering, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907 USA
| | - Jingwu Xie
- Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, 1040 W. Walnut Street., Indianapolis, IN, 46202, USA. .,Basic and Translational Cancer Review Branch (BTC), Division of Basic and Integrative Biological Sciences (DBIB), Center for Scientific Review, National Institutes of Health, 6701 Rockledge Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
461
|
Peperstraete K, Baes M, Swinkels D. Unexpected failure of rod bipolar cell targeting using L7Cre-2 mice. Exp Eye Res 2023; 228:109406. [PMID: 36740160 DOI: 10.1016/j.exer.2023.109406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Utilizing cell type-specific knockout mice has been an excellent tool for decades not only to explore the role of a gene in a specific cell, but also to unravel the underlying mechanism in diseases. To investigate the mechanistic association between dysfunction of the peroxisomal protein multifunctional protein 2 (MFP2) and retinopathy, we generated and phenotyped multiple transgenic mouse models with global or cell type-specific MFP2 deletion. These studies pointed to a potential role of MFP2 specifically in rod bipolar cells. To explore this, we aimed to create rod bipolar cell specific knockout mice of Mfp2 by crossing Mfp2L/L mice with L7Cre-2 mice (also known as PCP2Cre), generating L7-Mfp2-/- mice. L7Cre-2 mice express Cre recombinase under the control of the L7 promoter, which is believed to be exclusively expressed in rod bipolar cells and cerebellar Purkinje cells. Unexpectedly, only sporadic Cre activity was observed in the rod bipolar cells of L7-Mfp2-/- mice, despite efficient Cre recombination in cerebellar Purkinje cells. Moreover, a variable fraction of photoreceptors was targeted, which does not correspond with the supposed specificity of L7Cre-2 mice. These observations indicate that L7Cre-2 mice can be exploited to manipulate Purkinje cells in the cerebellum, whereas they cannot be used to generate rod bipolar cell specific knockout mice. For this aim, we suggest utilizing an independently generated mouse line named BAC-L7-IRES-Cre.
Collapse
Affiliation(s)
- Kaat Peperstraete
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| | - Daniëlle Swinkels
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
462
|
Henriet E, Knutsdottir H, Grasset EM, Dunworth M, Haynes M, Bader JS, Ewald AJ. Triple negative breast tumors contain heterogeneous cancer cells expressing distinct KRAS-dependent collective and disseminative invasion programs. Oncogene 2023; 42:737-747. [PMID: 36604566 PMCID: PMC10760065 DOI: 10.1038/s41388-022-02586-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023]
Abstract
Inter-patient and intra-tumoral heterogeneity complicate the identification of predictive biomarkers and effective treatments for basal triple negative breast cancer (b-TNBC). Invasion is the initiating event in metastasis and can occur by both collective and single-cell mechanisms. We cultured primary organoids from a b-TNBC genetically engineered mouse model in 3D collagen gels to characterize their invasive behavior. We observed that organoids from the same tumor presented different phenotypes that we classified as non-invasive, collective and disseminative. To identify molecular regulators driving these invasive phenotypes, we developed a workflow to isolate individual organoids from the collagen gels based on invasive morphology and perform RNA sequencing. We next tested the requirement of differentially regulated genes for invasion using shRNA knock-down. Strikingly, KRAS was required for both collective and disseminative phenotypes. We then performed a drug screen targeting signaling nodes upstream and downstream of KRAS. We found that inhibition of EGFR, MAPK/ERK, or PI3K/AKT signaling reduced invasion. Of these, ERK inhibition was striking for its ability to potently inhibit collective invasion and dissemination. We conclude that different cancer cells in the same b-TNBC tumor can express different metastatic molecular programs and identified KRAS and ERK as essential regulators of collective and single cell dissemination.
Collapse
Affiliation(s)
- Elodie Henriet
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hildur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Eloise M Grasset
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Matthew Dunworth
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Meagan Haynes
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Joel S Bader
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew J Ewald
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
463
|
Evano B, Sarde L, Tajbakhsh S. Temporal static and dynamic imaging of skeletal muscle in vivo. Exp Cell Res 2023; 424:113484. [PMID: 36693490 DOI: 10.1016/j.yexcr.2023.113484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
A major challenge in the study of living systems is understanding how tissues and organs are established, maintained during homeostasis, reconstituted following injury or deteriorated during disease. Most of the studies that interrogate in vivo cell biological properties of cell populations within tissues are obtained through static imaging approaches. However, in vertebrates, little is known about which, when, and how extracellular and intracellular signals are dynamically integrated to regulate cell behaviour and fates, due largely to technical challenges. Intravital imaging of cellular dynamics in mammalian models has exposed surprising properties that have been missed by conventional static imaging approaches. Here we highlight some selected examples of intravital imaging in mouse intestinal stem cells, hematopoietic stem cells, hair follicle stem cells, and neural stem cells in the brain, each of which have distinct features from an anatomical and niche-architecture perspective. Intravital imaging of mouse skeletal muscles is comparatively less advanced due to several technical constraints that will be discussed, yet this approach holds great promise as a complementary investigative method to validate findings obtained by static imaging, as well as a method for discovery.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, 75015, France; CNRS UMR 3738, Institut Pasteur, Paris, 75015, France
| | - Liza Sarde
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, 75015, France; CNRS UMR 3738, Institut Pasteur, Paris, 75015, France; Sorbonne Université, Complexité Du Vivant, F-75005, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, 75015, France; CNRS UMR 3738, Institut Pasteur, Paris, 75015, France.
| |
Collapse
|
464
|
Khadhraoui N, Prola A, Vandestienne A, Blondelle J, Guillaud L, Courtin G, Bodak M, Prost B, Huet H, Wintrebert M, Péchoux C, Solgadi A, Relaix F, Tiret L, Pilot-Storck F. Hacd2 deficiency in mice leads to an early and lethal mitochondrial disease. Mol Metab 2023; 69:101677. [PMID: 36693621 PMCID: PMC9986742 DOI: 10.1016/j.molmet.2023.101677] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Mitochondria fuel most animal cells with ATP, ensuring proper energetic metabolism of organs. Early and extensive mitochondrial dysfunction often leads to severe disorders through multiorgan failure. Hacd2 gene encodes an enzyme involved in very long chain fatty acid (C ≥ 18) synthesis, yet its roles in vivo remain poorly understood. Since mitochondria function relies on specific properties of their membranes conferred by a particular phospholipid composition, we investigated if Hacd2 gene participates to mitochondrial integrity. METHODS We generated two mouse models, the first one leading to a partial knockdown of Hacd2 expression and the second one, to a complete knockout of Hacd2 expression. We performed an in-depth analysis of the associated phenotypes, from whole organism to molecular scale. RESULTS Thanks to these models, we show that Hacd2 displays an early and broad expression, and that its deficiency in mice is lethal. Specifically, partial knockdown of Hacd2 expression leads to death within one to four weeks after birth, from a sudden growth arrest followed by cachexia and lethargy. The total knockout of Hacd2 is even more severe, characterized by embryonic lethality around E9.5 following developmental arrest and pronounced cardiovascular malformations. In-depth mechanistic analysis revealed that Hacd2 deficiency causes altered mitochondrial efficiency and ultrastructure, as well as accumulation of oxidized cardiolipin. CONCLUSIONS Altogether, these data indicate that the Hacd2 gene is essential for energetic metabolism during embryonic and postnatal development, acting through the control of proper mitochondrial organization and function.
Collapse
Affiliation(s)
- Nahed Khadhraoui
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Alexandre Prola
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Aymeline Vandestienne
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Jordan Blondelle
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Laurent Guillaud
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Guillaume Courtin
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Maxime Bodak
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Bastien Prost
- UMS IPSIT, Université Paris-Saclay, Châtenay-Malabry, F-92296, France
| | - Hélène Huet
- Biopôle, École nationale vétérinaire d'Alfort, Maisons-Alfort, F-94700, France
| | - Mélody Wintrebert
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Christine Péchoux
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, F-78350, Jouy-en-Josas, France
| | - Audrey Solgadi
- UMS IPSIT, Université Paris-Saclay, Châtenay-Malabry, F-92296, France
| | - Frédéric Relaix
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France
| | - Laurent Tiret
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France.
| | - Fanny Pilot-Storck
- Univ Paris-Est Créteil, INSERM, IMRB, Team Relaix, F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; EFS, IMRB, F-94010 Créteil, France.
| |
Collapse
|
465
|
Chiang IKN, Graus MS, Kirschnick N, Davidson T, Luu W, Harwood R, Jiang K, Li B, Wong YY, Moustaqil M, Lesieur E, Skoczylas R, Kouskoff V, Kazenwadel J, Arriola‐Martinez L, Sierecki E, Gambin Y, Alitalo K, Kiefer F, Harvey NL, Francois M. The blood vasculature instructs lymphatic patterning in a SOX7-dependent manner. EMBO J 2023; 42:e109032. [PMID: 36715213 PMCID: PMC9975944 DOI: 10.15252/embj.2021109032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/31/2023] Open
Abstract
Despite a growing catalog of secreted factors critical for lymphatic network assembly, little is known about the mechanisms that modulate the expression level of these molecular cues in blood vascular endothelial cells (BECs). Here, we show that a BEC-specific transcription factor, SOX7, plays a crucial role in a non-cell-autonomous manner by modulating the transcription of angiocrine signals to pattern lymphatic vessels. While SOX7 is not expressed in lymphatic endothelial cells (LECs), the conditional loss of SOX7 function in mouse embryos causes a dysmorphic dermal lymphatic phenotype. We identify novel distant regulatory regions in mice and humans that contribute to directly repressing the transcription of a major lymphangiogenic growth factor (Vegfc) in a SOX7-dependent manner. Further, we show that SOX7 directly binds HEY1, a canonical repressor of the Notch pathway, suggesting that transcriptional repression may also be modulated by the recruitment of this protein partner at Vegfc genomic regulatory regions. Our work unveils a role for SOX7 in modulating downstream signaling events crucial for lymphatic patterning, at least in part via the transcriptional repression of VEGFC levels in the blood vascular endothelium.
Collapse
Affiliation(s)
- Ivy K N Chiang
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Matthew S Graus
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Nils Kirschnick
- European Institute for Molecular Imaging (EIMI)University of MünsterMünsterGermany
| | - Tara Davidson
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Winnie Luu
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Richard Harwood
- Sydney Microscopy and MicroanalysisUniversity of SydneySydneyNSWAustralia
| | - Keyi Jiang
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Bitong Li
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Yew Yan Wong
- The Genome Imaging CenterThe Centenary InstituteSydneyNSWAustralia
| | - Mehdi Moustaqil
- EMBL Australia Node in Single Molecule Science, and School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Emmanuelle Lesieur
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaQLDAustralia
| | - Renae Skoczylas
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaQLDAustralia
| | - Valerie Kouskoff
- Division of Developmental Biology & MedicineThe University of ManchesterManchesterUK
| | - Jan Kazenwadel
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSAAustralia
| | - Luis Arriola‐Martinez
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSAAustralia
| | - Emma Sierecki
- EMBL Australia Node in Single Molecule Science, and School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Yann Gambin
- EMBL Australia Node in Single Molecule Science, and School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Friedmann Kiefer
- European Institute for Molecular Imaging (EIMI)University of MünsterMünsterGermany
| | - Natasha L Harvey
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSAAustralia
| | - Mathias Francois
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
- The Genome Imaging CenterThe Centenary InstituteSydneyNSWAustralia
| |
Collapse
|
466
|
Mohamed GA, Mahmood S, Ognjenovic NB, Lee MK, Wilkins OM, Christensen BC, Muller KE, Pattabiraman DR. Lineage plasticity enables low-ER luminal tumors to evolve and gain basal-like traits. Breast Cancer Res 2023; 25:23. [PMID: 36859337 PMCID: PMC9979432 DOI: 10.1186/s13058-023-01621-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
Stratifying breast cancer into specific molecular or histologic subtypes aids in therapeutic decision-making and predicting outcomes; however, these subtypes may not be as distinct as previously thought. Patients with luminal-like, estrogen receptor (ER)-expressing tumors have better prognosis than patients with more aggressive, triple-negative or basal-like tumors. There is, however, a subset of luminal-like tumors that express lower levels of ER, which exhibit more basal-like features. We have found that breast tumors expressing lower levels of ER, traditionally considered to be luminal-like, represent a distinct subset of breast cancer characterized by the emergence of basal-like features. Lineage tracing of low-ER tumors in the MMTV-PyMT mouse mammary tumor model revealed that basal marker-expressing cells arose from normal luminal epithelial cells, suggesting that luminal-to-basal plasticity is responsible for the evolution and emergence of basal-like characteristics. This plasticity allows tumor cells to gain a new lumino-basal phenotype, thus leading to intratumoral lumino-basal heterogeneity. Single-cell RNA sequencing revealed SOX10 as a potential driver for this plasticity, which is known among breast tumors to be almost exclusively expressed in triple-negative breast cancer (TNBC) and was also found to be highly expressed in low-ER tumors. These findings suggest that basal-like tumors may result from the evolutionary progression of luminal tumors with low ER expression.
Collapse
Affiliation(s)
- Gadisti Aisha Mohamed
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Sundis Mahmood
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Nevena B Ognjenovic
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Min Kyung Lee
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Owen M Wilkins
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Brock C Christensen
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Kristen E Muller
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA.
| | - Diwakar R Pattabiraman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA.
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA.
| |
Collapse
|
467
|
Moayedi Y, Xu S, Obayashi SK, Hoffman BU, Gerling GJ, Lumpkin EA. The cellular basis of mechanosensation in mammalian tongue. Cell Rep 2023; 42:112087. [PMID: 36763499 PMCID: PMC10409885 DOI: 10.1016/j.celrep.2023.112087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Mechanosensory neurons that innervate the tongue provide essential information to guide feeding, speech, and social grooming. We use in vivo calcium imaging of mouse trigeminal ganglion neurons to identify functional groups of mechanosensory neurons innervating the anterior tongue. These sensory neurons respond to thermal and mechanical stimulation. Analysis of neuronal activity patterns reveal that most mechanosensory trigeminal neurons are tuned to detect moving stimuli across the tongue. Using an unbiased, multilayer hierarchical clustering approach to classify pressure-evoked activity based on temporal response dynamics, we identify five functional classes of mechanosensory neurons with distinct force-response relations and adaptation profiles. These populations are tuned to detect different features of touch. Molecular markers of functionally distinct clusters are identified by analyzing cluster representation in genetically marked neuronal subsets. Collectively, these studies provide a platform for defining the contributions of functionally distinct mechanosensory neurons to oral behaviors crucial for survival in mammals.
Collapse
Affiliation(s)
- Yalda Moayedi
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA; Department of Otolaryngology - Head & Neck Surgery, Columbia University, New York, NY 10032, USA
| | - Shan Xu
- School of Engineering and Applied Science, University of Virginia, Charlottesville, VA 22904, USA
| | - Sophie K Obayashi
- Department of Molecular & Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Benjamin U Hoffman
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Gregory J Gerling
- School of Engineering and Applied Science, University of Virginia, Charlottesville, VA 22904, USA.
| | - Ellen A Lumpkin
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA; Department of Molecular & Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
468
|
Manieri E, Tie G, Seruggia D, Madha S, Maglieri A, Huang K, Fujiwara Y, Zhang K, Orkin SH, He R, McCarthy N, Shivdasani RA. Defining the structure, signals, and cellular elements of the gastric mesenchymal niche. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.11.527728. [PMID: 36798304 PMCID: PMC9934611 DOI: 10.1101/2023.02.11.527728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
PDGFRA-expressing mesenchyme provides a niche for intestinal stem cells. Corresponding compartments are unknown in the stomach, where corpus and antral glandular epithelia have similar niche dependencies but are structurally distinct from the intestine and from each other. Previous studies considered antrum and corpus as a whole and did not assess niche functions. Using high-resolution imaging and sequencing, we identify regional subpopulations and niche properties of purified mouse corpus and antral PDGFRA + cells. PDGFRA Hi sub-epithelial myofibroblasts are principal sources of BMP ligands in both gastric segments; two molecularly distinct groups distribute asymmetrically along antral glands but together fail to support epithelial organoids in vitro . In contrast, strategically positioned PDGFRA Lo cells that express CD55 enable corpus and antral organoid growth in the absence of other cellular or soluble factors. Our study provides detailed insights into spatial, molecular, and functional organization of gastric mesenchyme and the spectrum of signaling sources for stem cell support.
Collapse
|
469
|
Greig LC, Woodworth MB, Poulopoulos A, Lim S, Macklis JD. BEAM: a combinatorial recombinase toolbox for binary gene expression and mosaic analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.16.528875. [PMID: 36824714 PMCID: PMC9949094 DOI: 10.1101/2023.02.16.528875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Genetic mosaic analysis, in which mutant cells reside intermingled with wild-type cells, is a powerful experimental approach, but has not been widely used in mice because existing genome-based strategies require complicated and protracted breeding schemes. We have developed an alternative approach termed BEAM (for Binary Expression Aleatory Mosaic) that relies on sparse recombinase activation to generate two genetically distinct, non-overlapping populations of cells for comparative analysis. Following delivery of DNA constructs by transfection or viral transduction, combinatorial recombinase activity generates two distinct populations of cells labeled with either green or red fluorescent protein. Any gene of interest can be mis-expressed or deleted in one population for comparison with intermingled control cells. We have extensively optimized and characterized this system both in vitro and in vivo , and demonstrate its power for investigating cell autonomy, identifying temporally or spatially aberrant phenotypes, revealing changes in cell proliferation or death, and controlling for procedural variability.
Collapse
|
470
|
George RM, Firulli BA, Podicheti R, Rusch DB, Mannion BJ, Pennacchio LA, Osterwalder M, Firulli AB. Single cell evaluation of endocardial Hand2 gene regulatory networks reveals HAND2-dependent pathways that impact cardiac morphogenesis. Development 2023; 150:dev201341. [PMID: 36620995 PMCID: PMC10110492 DOI: 10.1242/dev.201341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
The transcription factor HAND2 plays essential roles during cardiogenesis. Hand2 endocardial deletion (H2CKO) results in tricuspid atresia or double inlet left ventricle with accompanying intraventricular septum defects, hypo-trabeculated ventricles and an increased density of coronary lumens. To understand the regulatory mechanisms of these phenotypes, single cell transcriptome analysis of mouse E11.5 H2CKO hearts was performed revealing a number of disrupted endocardial regulatory pathways. Using HAND2 DNA occupancy data, we identify several HAND2-dependent enhancers, including two endothelial enhancers for the shear-stress master regulator KLF2. A 1.8 kb enhancer located 50 kb upstream of the Klf2 TSS imparts specific endothelial/endocardial expression within the vasculature and endocardium. This enhancer is HAND2-dependent for ventricular endocardium expression but HAND2-independent for Klf2 vascular and valve expression. Deletion of this Klf2 enhancer results in reduced Klf2 expression within ventricular endocardium. These data reveal that HAND2 functions within endocardial gene regulatory networks including shear-stress response.
Collapse
Affiliation(s)
- Rajani M. George
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Beth A. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Douglas B. Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Brandon J. Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
| | - Len A. Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern 3008, Switzerland
- Department of Cardiology, Bern University Hospital, Bern 3010, Switzerland
| | - Anthony B. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| |
Collapse
|
471
|
de Almeida MMA, Watson AES, Bibi S, Dittmann NL, Goodkey K, Sharafodinzadeh P, Galleguillos D, Nakhaei-Nejad M, Kosaraju J, Steinberg N, Wang BS, Footz T, Giuliani F, Wang J, Sipione S, Edgar JM, Voronova A. Fractalkine enhances oligodendrocyte regeneration and remyelination in a demyelination mouse model. Stem Cell Reports 2023; 18:519-533. [PMID: 36608690 PMCID: PMC9968989 DOI: 10.1016/j.stemcr.2022.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
Demyelinating disorders of the central nervous system (CNS) occur when myelin and oligodendrocytes are damaged or lost. Remyelination and regeneration of oligodendrocytes can be achieved from endogenous oligodendrocyte precursor cells (OPCs) that reside in the adult CNS tissue. Using a cuprizone mouse model of demyelination, we show that infusion of fractalkine (CX3CL1) into the demyelinated murine brain increases de novo oligodendrocyte formation and enhances remyelination in the corpus callosum and cortical gray matter. This is achieved by increased OPC proliferation in the cortical gray matter as well as OPC differentiation and attenuation of microglia/macrophage activation both in corpus callosum and cortical gray matter. Finally, we show that activated OPCs and microglia/macrophages express fractalkine receptor CX3CR1 in vivo, and that in OPC-microglia co-cultures fractalkine increases in vitro oligodendrocyte differentiation by modulating both OPC and microglia biology. Our results demonstrate a novel pro-regenerative role of fractalkine in a demyelinating mouse model.
Collapse
Affiliation(s)
- Monique M A de Almeida
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada
| | - Sana Bibi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada
| | - Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada
| | - Pedram Sharafodinzadeh
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Danny Galleguillos
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Maryam Nakhaei-Nejad
- Department of Medicine, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Jayasankar Kosaraju
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Noam Steinberg
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Beatrix S Wang
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada
| | - Tim Footz
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Fabrizio Giuliani
- Department of Medicine, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada; Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Jing Wang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Simonetta Sipione
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Julia M Edgar
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada; Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada; Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
472
|
Cheng Q, Wu J, Xia Y, Cheng Q, Zhao Y, Zhu P, Zhang W, Zhang S, Zhang L, Yuan Y, Li C, Chen G, Xue B. Disruption of protein geranylgeranylation in the cerebellum causes cerebellar hypoplasia and ataxia via blocking granule cell progenitor proliferation. Mol Brain 2023; 16:24. [PMID: 36782228 PMCID: PMC9923931 DOI: 10.1186/s13041-023-01010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
The prenylation of proteins is involved in a variety of biological functions. However, it remains unknown whether it plays an important role in the morphogenesis of the cerebellum. To address this question, we generated a mouse model, in which the geranylgeranyl pyrophosphate synthase (Ggps1) gene is inactivated in neural progenitor cells in the developing cerebellum. We report that conditional knockout (cKO) of Ggps1 leads to severe ataxia and deficient locomotion. To identify the underlying mechanisms, we completed a series of cellular and molecular experiments. First, our morphological analysis revealed significantly decreased population of granule cell progenitors (GCPs) and impaired proliferation of GCPs in the developing cerebellum of Ggps1 cKO mice. Second, our molecular analysis showed increased expression of p21, an important cell cycle regulator in Ggps1 cKO mice. Together, this study highlights a critical role of Ggpps-dependent protein prenylation in the proliferation of cerebellar GCPs during cerebellar development.
Collapse
Affiliation(s)
- Qi Cheng
- grid.41156.370000 0001 2314 964XMedical School of Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093 China
| | - Jing Wu
- grid.89957.3a0000 0000 9255 8984Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166 China
| | - Yingqian Xia
- grid.41156.370000 0001 2314 964XMedical School of Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093 China
| | - Qing Cheng
- grid.89957.3a0000 0000 9255 8984Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004 Jiangsu China
| | - Yinjuan Zhao
- grid.410625.40000 0001 2293 4910Collaborative Innovation Center of Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 Jiangsu China
| | - Peixiang Zhu
- grid.41156.370000 0001 2314 964XMedical School of Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093 China
| | - Wangling Zhang
- grid.41156.370000 0001 2314 964XMedical School of Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093 China
| | - Shihu Zhang
- grid.410745.30000 0004 1765 1045Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029 China
| | - Lei Zhang
- Medical Imaging Center of Fuyang People’s Hospital, Fuyang, Anhui Province China
| | - Yushan Yuan
- Medical Imaging Center of Fuyang People’s Hospital, Fuyang, Anhui Province China
| | - Chaojun Li
- State Key Laboratory of Reproductive Medicine and China International Joint Research Center On Environment and Human Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Guiquan Chen
- Medical School of Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China. .,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
473
|
Navaridas R, Vidal-Sabanés M, Ruiz-Mitjana A, Perramon-Güell A, Megino-Luque C, Llobet-Navas D, Matias-Guiu X, Egea J, Encinas M, Bardia L, Colombelli J, Dolcet X. Transient and DNA-free in vivo CRISPR/Cas9 genome editing for flexible modeling of endometrial carcinogenesis. Cancer Commun (Lond) 2023; 43:620-624. [PMID: 36762520 PMCID: PMC10174088 DOI: 10.1002/cac2.12409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Affiliation(s)
- Raúl Navaridas
- Oncological Pathology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Maria Vidal-Sabanés
- Oncological Pathology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Anna Ruiz-Mitjana
- Oncological Pathology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Aida Perramon-Güell
- Oncological Pathology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Cristina Megino-Luque
- Oncological Pathology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - David Llobet-Navas
- Molecular Mechanisms and Experimental Therapy in Oncology-Oncobell Program, Bellvitge Biomedical Research Institute, 'L'Hospitalet de Llobregat, Barcelona, Spain.,Cancer Networking Biomedical Research Center, Institute of Health Carlos III, Madrid, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Bellvitge Hospital, University of Barcelona, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Pathology, Arnau de Vilanova Hospital, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain.,Cancer Networking Biomedical Research Center, Institute of Health Carlos III, Madrid, Spain
| | - Joaquim Egea
- Molecular Developmental Neurobiology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Mario Encinas
- Developmental and Oncogenic Signalling Group, Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Lídia Bardia
- Advanced Digital Microscopy Core Facility, Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Julien Colombelli
- Advanced Digital Microscopy Core Facility, Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Xavier Dolcet
- Oncological Pathology Group, Department of Basic Medical Sciences, University of Lleida, Biomedical Research Institute of Lleida, Lleida, Spain
| |
Collapse
|
474
|
Hu G, Song M, Wang Y, Hao K, Wang J, Zhang Y. Using a modified piggyBac transposon-combined Cre/loxP system to produce selectable reporter-free transgenic bovine mammary epithelial cells for somatic cell nuclear transfer. Genesis 2023:e23510. [PMID: 36748563 DOI: 10.1002/dvg.23510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 02/08/2023]
Abstract
Transposon systems are widely used for genetic engineering in various model organisms. PiggyBac (PB) has recently been confirmed to have highly efficient transposition in the mouse germ line and mammalian cell lines. In this study, we used a modified PB transposon system mediated by PB transposase (PBase) mRNA carrying the human lactoferrin gene driven by bovine β-casein promoter to transfect bovine mammary epithelial cells (BMECs), and the selectable reporter in two stable transgenic BMEC clones was removed using cell-permeant Cre recombinase. These reporter-free transgenic BMECs were used as donor cells for somatic cell nuclear transfer (SCNT) and exhibited a competence of SCNT embryos similar to stable transgenic BMECs and nontransgenic BMECs. The comprehensive information from this study provided a modified approach using an altered PB transposon system mediated by PBase mRNA in vitro and combined with the Cre/loxP system to produce transgenic and selectable reporter-free donor nuclei for SCNT. Consequently, the production of safe bovine mammary bioreactors can be promoted.
Collapse
Affiliation(s)
- Guangdong Hu
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Meijun Song
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yan Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Kexing Hao
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jing Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
475
|
Castillo-Azofeifa D, Wald T, Reyes EA, Gallagher A, Schanin J, Vlachos S, Lamarche-Vane N, Bomidi C, Blutt S, Estes MK, Nystul T, Klein OD. A DLG1-ARHGAP31-CDC42 axis is essential for the intestinal stem cell response to fluctuating niche Wnt signaling. Cell Stem Cell 2023; 30:188-206.e6. [PMID: 36640764 PMCID: PMC9922544 DOI: 10.1016/j.stem.2022.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/13/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023]
Abstract
A central factor in the maintenance of tissue integrity is the response of stem cells to variations in the levels of niche signals. In the gut, intestinal stem cells (ISCs) depend on Wnt ligands for self-renewal and proliferation. Transient increases in Wnt signaling promote regeneration after injury or in inflammatory bowel diseases, whereas constitutive activation of this pathway leads to colorectal cancer. Here, we report that Discs large 1 (Dlg1), although dispensable for polarity and cellular turnover during intestinal homeostasis, is required for ISC survival in the context of increased Wnt signaling. RNA sequencing (RNA-seq) and genetic mouse models demonstrated that DLG1 regulates the cellular response to increased canonical Wnt ligands. This occurs via the transcriptional regulation of Arhgap31, a GTPase-activating protein that deactivates CDC42, an effector of the non-canonical Wnt pathway. These findings reveal a DLG1-ARHGAP31-CDC42 axis that is essential for the ISC response to increased niche Wnt signaling.
Collapse
Affiliation(s)
- David Castillo-Azofeifa
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Regenerative Medicine, Genentech, Inc., South San Francisco, CA, USA
| | - Tomas Wald
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Efren A Reyes
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pharmaceutical Chemistry and TETRAD Program, University of California, San Francisco, San Francisco, CA, USA
| | - Aaron Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Schanin
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie Vlachos
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sarah Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Todd Nystul
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
476
|
Landstrom AP, Yang Q, Sun B, Perelli RM, Bidzimou MT, Zhang Z, Aguilar-Sanchez Y, Alsina KM, Cao S, Reynolds JO, Word TA, van der Sangen NM, Wells Q, Kannankeril PJ, Ludwig A, Kim JJ, Wehrens XH. Reduction in Junctophilin 2 Expression in Cardiac Nodal Tissue Results in Intracellular Calcium-Driven Increase in Nodal Cell Automaticity. Circ Arrhythm Electrophysiol 2023; 16:e010858. [PMID: 36706317 PMCID: PMC9974897 DOI: 10.1161/circep.122.010858] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/06/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Spontaneously depolarizing nodal cells comprise the pacemaker of the heart. Intracellular calcium (Ca2+) plays a critical role in mediating nodal cell automaticity and understanding this so-called Ca2+ clock is critical to understanding nodal arrhythmias. We previously demonstrated a role for Jph2 (junctophilin 2) in regulating Ca2+-signaling through inhibition of RyR2 (ryanodine receptor 2) Ca2+ leak in cardiac myocytes; however, its role in pacemaker function and nodal arrhythmias remains unknown. We sought to determine whether nodal Jph2 expression silencing causes increased sinoatrial and atrioventricular nodal cell automaticity due to aberrant RyR2 Ca2+ leak. METHODS A tamoxifen-inducible, nodal tissue-specific, knockdown mouse of Jph2 was achieved using a Cre-recombinase-triggered short RNA hairpin directed against Jph2 (Hcn4:shJph2). In vivo cardiac rhythm was monitored by surface ECG, implantable cardiac telemetry, and intracardiac electrophysiology studies. Intracellular Ca2+ imaging was performed using confocal-based line scans of isolated nodal cells loaded with fluorescent Ca2+ reporter Cal-520. Whole cell patch clamp was conducted on isolated nodal cells to determine action potential kinetics and sodium-calcium exchanger function. RESULTS Hcn4:shJph2 mice demonstrated a 40% reduction in nodal Jph2 expression, resting sinus tachycardia, and impaired heart rate response to pharmacologic stress. In vivo intracardiac electrophysiology studies and ex vivo optical mapping demonstrated accelerated junctional rhythm originating from the atrioventricular node. Hcn4:shJph2 nodal cells demonstrated increased and irregular Ca2+ transient generation with increased Ca2+ spark frequency and Ca2+ leak from the sarcoplasmic reticulum. This was associated with increased nodal cell AP firing rate, faster diastolic repolarization rate, and reduced sodium-calcium exchanger activity during repolarized states compared to control. Phenome-wide association studies of the JPH2 locus identified an association with sinoatrial nodal disease and atrioventricular nodal block. CONCLUSIONS Nodal-specific Jph2 knockdown causes increased nodal automaticity through increased Ca2+ leak from intracellular stores. Dysregulated intracellular Ca2+ underlies nodal arrhythmogenesis in this mouse model.
Collapse
Affiliation(s)
- Andrew P. Landstrom
- Dept of Pediatrics, Division of Cardiology, Duke Univ School of Medicine, Durham, NC
- Dept of Cell Biology, Duke Univ School of Medicine, Durham, NC
| | - Qixin Yang
- Dept of Pediatrics, Division of Cardiology, Duke Univ School of Medicine, Durham, NC
- Dept of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang Univ, Hangzhou, China
| | - Bo Sun
- Dept of Pediatrics, Division of Cardiology, Duke Univ School of Medicine, Durham, NC
| | | | | | - Zhushan Zhang
- Dept of Cell Biology, Duke Univ School of Medicine, Durham, NC
| | - Yuriana Aguilar-Sanchez
- Integrative Molecular & Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | - Katherina M. Alsina
- Integrative Molecular & Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | - Shuyi Cao
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| | - Julia O. Reynolds
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| | - Tarah A. Word
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| | | | - Quinn Wells
- Depts of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Prince J. Kannankeril
- Center for Pediatric Precision Medicine, Dept of Pediatrics, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jeffrey J. Kim
- Dept of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX
| | - Xander H.T. Wehrens
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
- Dept of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX
- Depts of Neuroscience & Center for Space Medicine and the Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
| |
Collapse
|
477
|
Adamova P, Lotto RR, Powell AK, Dykes IM. Are there foetal extracellular vesicles in maternal blood? Prospects for diagnostic biomarker discovery. J Mol Med (Berl) 2023; 101:65-81. [PMID: 36538060 PMCID: PMC9977902 DOI: 10.1007/s00109-022-02278-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 03/02/2023]
Abstract
Prenatal diagnosis of congenital disease improves clinical outcomes; however, as many as 50% of congenital heart disease cases are missed by current ultrasound screening methods. This indicates a need for improved screening technology. Extracellular vesicles (EVs) have attracted enormous interest in recent years for their potential in diagnostics. EVs mediate endocrine signalling in health and disease and are known to regulate aspects of embryonic development. Here, we critically evaluate recent evidence suggesting that EVs released from the foetus are able to cross the placenta and enter the maternal circulation. Furthermore, EVs from the mother appear to be transported in the reverse direction, whilst the placenta itself acts as a source of EVs. Experimental work utilising rodent models employing either transgenically encoded reporters or application of fluorescent tracking dyes provide convincing evidence of foetal-maternal crosstalk. This is supported by clinical data demonstrating expression of placental-origin EVs in maternal blood, as well as limited evidence for the presence of foetal-origin EVs. Together, this work raises the possibility that foetal EVs present in maternal blood could be used for the diagnosis of congenital disease. We discuss the challenges faced by researchers in translating these basic science findings into a clinical non-invasive prenatal test.
Collapse
Affiliation(s)
- Petra Adamova
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK.,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK
| | - Robyn R Lotto
- Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK.,School of Nursing and Allied Health, Liverpool John Moores University, Tithebarn St, Liverpool, L2 2ER, UK
| | - Andrew K Powell
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK.,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK
| | - Iain M Dykes
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK. .,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
478
|
Loughlin S, Costello HM, Roe AJ, Buckley C, Wilson SM, Bailey MA, Mansley MK. Mapping the Transcriptome Underpinning Acute Corticosteroid Action within the Cortical Collecting Duct. KIDNEY360 2023; 4:226-240. [PMID: 36821614 PMCID: PMC10103384 DOI: 10.34067/kid.0003582022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2022]
Abstract
Key Points We report the transcriptomes associated with acute corticosteroid regulation of ENaC activity in polarized mCCDcl1 collecting duct cells. Nine genes were regulated by aldosterone (ALDO), 0 with corticosterone alone, and 151 with corticosterone when 11βHSD2 activity was inhibited. We validated three novel ALDO-induced genes, Rasd1 , Sult1d1 , and Gm43305 , in primary cells isolated from a novel principal cell reporter mouse. Background Corticosteroids regulate distal nephron and collecting duct (CD) Na+ reabsorption, contributing to fluid-volume and blood pressure homeostasis. The transcriptional landscape underpinning the acute stimulation of the epithelial sodium channel (ENaC) by physiological concentrations of corticosteroids remains unclear. Methods Transcriptomic profiles underlying corticosteroid-stimulated ENaC activity in polarized mCCDcl1 cells were generated by coupling electrophysiological measurements of amiloride-sensitive currents with RNAseq. Generation of a principal cell-specific reporter mouse line, mT/mG -Aqp2Cre, enabled isolation of primary CD principal cells by FACS, and ENaC activity was measured in cultured primary cells after acute application of corticosteroids. Expression of target genes was assessed by qRT-PCR in cultured cells or freshly isolated cells after the acute elevation of steroid hormones in mT/mG -Aqp2Cre mice. Results Physiological relevance of the mCCDcl1 model was confirmed with aldosterone (ALDO)-specific stimulation of SGK1 and ENaC activity. Corticosterone (CORT) only modulated these responses at supraphysiological concentrations or when 11βHSD2 was inhibited. When 11βHSD2 protection was intact, CORT caused no significant change in transcripts. We identified a small number of ALDO-induced transcripts associated with stimulated ENaC activity in mCCDcl1 cells and a much larger number with CORT in the absence of 11βHSD2 activity. Principal cells isolated from mT/mG -Aqp2Cre mice were validated and assessment of identified ALDO-induced genes revealed that Sgk1 , Zbtbt16 , Sult1d1 , Rasd1 , and Gm43305 are acutely upregulated by corticosteroids both in vitro and in vivo . Conclusions This study reports the transcriptome of mCCDcl1 cells and identifies a small number of ALDO-induced genes associated with acute stimulation of ENaC, including three previously undescribed genes.
Collapse
Affiliation(s)
- Struan Loughlin
- Cellular Medicine Research Division, University of St Andrews, St Andrews, United Kingdom
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hannah M. Costello
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J. Roe
- Division of Pharmacy, School of Medicine, Pharmacy and Health, Durham University Queen's Campus, Stockton-on-Tees, United Kingdom
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Stuart M. Wilson
- Division of Pharmacy, School of Medicine, Pharmacy and Health, Durham University Queen's Campus, Stockton-on-Tees, United Kingdom
| | - Matthew A. Bailey
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Morag K. Mansley
- Cellular Medicine Research Division, University of St Andrews, St Andrews, United Kingdom
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Division of Pharmacy, School of Medicine, Pharmacy and Health, Durham University Queen's Campus, Stockton-on-Tees, United Kingdom
| |
Collapse
|
479
|
Min KW, Kim N, Lee JH, Sung Y, Kim M, Lee EJ, Kim JM, Kim JH, Lee J, Cho W, Yang JM, Kim N, Kim J, Lee CJ, Park YG, Lee SH, Lee HW, Kim JW. Visuomotor anomalies in achiasmatic mice expressing a transfer-defective Vax1 mutant. Exp Mol Med 2023; 55:385-400. [PMID: 36737666 PMCID: PMC9981622 DOI: 10.1038/s12276-023-00930-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 02/05/2023] Open
Abstract
In binocular animals that exhibit stereoscopic visual responses, the axons of retinal ganglion cells (RGCs) connect to brain areas bilaterally by forming a commissure called the optic chiasm (OC). Ventral anterior homeobox 1 (Vax1) contributes to the formation of the OC, acting endogenously in optic pathway cells and exogenously in growing RGC axons. Here, we generated Vax1AA/AA mice expressing the Vax1AA mutant, which is incapable of intercellular transfer. We found that RGC axons cannot take up Vax1AA protein from the Vax1AA/AA mouse optic stalk (OS) and grow slowly to arrive at the hypothalamus at a late stage. The RGC axons of Vax1AA/AA mice connect exclusively to ipsilateral brain areas after failing to access the midline, resulting in reduced visual acuity and abnormal oculomotor responses. Overall, our study provides physiological evidence for the necessity of intercellular transfer of Vax1 and the importance of the bilateral RGC axon projection in proper visuomotor responses.
Collapse
Affiliation(s)
- Kwang Wook Min
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Namsuk Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.,Neurovascular Unit, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jae Hoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Younghoon Sung
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Museong Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Eun Jung Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jong-Myeong Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jae-Hyun Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jaeyoung Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Wonjin Cho
- Department of Bio & Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jee Myung Yang
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Dongguk-ro 27, Ilsandong-gu, Goyang, Gyeong-gi, Republic of Korea
| | - Nury Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Jaehoon Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Young-Gyun Park
- Department of Bio & Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seung-Hee Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jin Woo Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
480
|
Ampartzidis I, Efstathiou C, Paonessa F, Thompson EM, Wilson T, McCann CJ, Greene NDE, Copp AJ, Livesey FJ, Elvassore N, Giobbe GG, De Coppi P, Maniou E, Galea GL. Synchronisation of apical constriction and cell cycle progression is a conserved behaviour of pseudostratified neuroepithelia informed by their tissue geometry. Dev Biol 2023; 494:60-70. [PMID: 36509125 PMCID: PMC10570144 DOI: 10.1016/j.ydbio.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
Neuroepithelial cells balance tissue growth requirement with the morphogenetic imperative of closing the neural tube. They apically constrict to generate mechanical forces which elevate the neural folds, but are thought to apically dilate during mitosis. However, we previously reported that mitotic neuroepithelial cells in the mouse posterior neuropore have smaller apical surfaces than non-mitotic cells. Here, we document progressive apical enrichment of non-muscle myosin-II in mitotic, but not non-mitotic, neuroepithelial cells with smaller apical areas. Live-imaging of the chick posterior neuropore confirms apical constriction synchronised with mitosis, reaching maximal constriction by anaphase, before division and re-dilation. Mitotic apical constriction amplitude is significantly greater than interphase constrictions. To investigate conservation in humans, we characterised early stages of iPSC differentiation through dual SMAD-inhibition to robustly produce pseudostratified neuroepithelia with apically enriched actomyosin. These cultured neuroepithelial cells achieve an equivalent apical area to those in mouse embryos. iPSC-derived neuroepithelial cells have large apical areas in G2 which constrict in M phase and retain this constriction in G1/S. Given that this differentiation method produces anterior neural identities, we studied the anterior neuroepithelium of the elevating mouse mid-brain neural tube. Instead of constricting, mid-brain mitotic neuroepithelial cells have larger apical areas than interphase cells. Tissue geometry differs between the apically convex early midbrain and flat posterior neuropore. Culturing human neuroepithelia on equivalently convex surfaces prevents mitotic apical constriction. Thus, neuroepithelial cells undergo high-amplitude apical constriction synchronised with cell cycle progression but the timing of their constriction if influenced by tissue geometry.
Collapse
Affiliation(s)
- Ioakeim Ampartzidis
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Christoforos Efstathiou
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Francesco Paonessa
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Elliott M Thompson
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Tyler Wilson
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Conor J McCann
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Nicholas DE Greene
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Andrew J Copp
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Frederick J Livesey
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Nicola Elvassore
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; Veneto Institute of Molecular Medicine, Padova, Italy; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Giovanni G Giobbe
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Paolo De Coppi
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK; Specialist Neonatal and Paediatric Unit, Great Ormond Street Hospital, London, WC1N 1EH, UK
| | - Eirini Maniou
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gabriel L Galea
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
481
|
Ayala-Sarmiento AE, Pacheco DRF, Breunig JJ. Murine-Derived Glioma Organoids and Cell Line Culture Systems. Curr Protoc 2023; 3:e665. [PMID: 36744986 PMCID: PMC9909623 DOI: 10.1002/cpz1.665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Research models in cancer have greatly evolved in the last decade, with the advent of several new methods both in vitro and in vivo. While in vivo models remain the gold standard for preclinical studies, these methods present a series of disadvantages such as a high cost and long periods of time to produce results compared with in vitro models. We have previously developed a method named Mosaic Analysis by Dual Recombinase-mediated cassette exchange (MADR) that generates autochthonous gliomas in immunocompetent mice through the transgenesis of personalized driver mutations, which highly mimic the spatial and temporal tumor development of their human counterparts. Due to the control of single-copy expression of transgenes, it allows for comparing the visualization of tumor cells and non-tumor cells. Here we describe a method to generate murine-derived glioma organoids (MGOs) and cell line cultures from these murine models by physical and enzymatic methods for in vitro downstream applications. Tumor cells can be readily distinguished from non-tumor cell populations, in both organoids and monolayer cell cultures, and isolated due to the use of personalized fluorescent reporter transgenes. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Generation of 3D murine-derived glioma organoids Basic Protocol 2: Generation of 2D glioma monolayer cell lines.
Collapse
Affiliation(s)
- Alberto E. Ayala-Sarmiento
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- These authors contributed equally
| | - David Rincon Fernandez Pacheco
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- These authors contributed equally
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
482
|
Sarkar A, Jin Y, DeFelice BC, Logan CY, Yang Y, Anbarchian T, Wu P, Morri M, Neff NF, Nguyen H, Rulifson E, Fish M, Kaye AG, Martínez Jaimes AM, Nusse R. Intermittent fasting induces rapid hepatocyte proliferation to restore the hepatostat in the mouse liver. eLife 2023; 12:e82311. [PMID: 36719070 PMCID: PMC9889086 DOI: 10.7554/elife.82311] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/09/2022] [Indexed: 02/01/2023] Open
Abstract
Nutrient availability fluctuates in most natural populations, forcing organisms to undergo periods of fasting and re-feeding. It is unknown how dietary changes influence liver homeostasis. Here, we show that a switch from ad libitum feeding to intermittent fasting (IF) promotes rapid hepatocyte proliferation. Mechanistically, IF-induced hepatocyte proliferation is driven by the combined action of systemic FGF15 and localized WNT signaling. Hepatocyte proliferation during periods of fasting and re-feeding re-establishes a constant liver-to-body mass ratio, thus maintaining the hepatostat. This study provides the first example of dietary influence on adult hepatocyte proliferation and challenges the widely held view that liver tissue is mostly quiescent unless chemically or mechanically injured.
Collapse
Affiliation(s)
- Abby Sarkar
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yinhua Jin
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | | | - Catriona Y Logan
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yan Yang
- Stanford Center for Genomics & Personalized Medicine, Stanford University School of MedicineStanfordUnited States
| | - Teni Anbarchian
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Peng Wu
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | | | - Norma F Neff
- Chan-Zuckerberg BiohubSan FranciscoUnited States
| | - Huy Nguyen
- Department of Neurology and Neurological Sciences, Stanford University School of MedicineStanfordUnited States
| | - Eric Rulifson
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Matthew Fish
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Avi Gurion Kaye
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Azalia M Martínez Jaimes
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Roel Nusse
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
483
|
Zou J, Wang W, Lu Y, Ayala J, Dong K, Zhou H, Wang J, Chen W, Weintraub NL, Zhou J, Li J, Su H. Neddylation is required for perinatal cardiac development through stimulation of metabolic maturation. Cell Rep 2023; 42:112018. [PMID: 36662623 PMCID: PMC10029150 DOI: 10.1016/j.celrep.2023.112018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Cardiac maturation is crucial for postnatal cardiac development and is increasingly known to be regulated by a series of transcription factors. However, post-translational mechanisms regulating this process remain unclear. Here we report the indispensable role of neddylation in cardiac maturation. Mosaic deletion of NAE1, an essential enzyme for neddylation, in neonatal hearts results in the rapid development of cardiomyopathy and heart failure. NAE1 deficiency disrupts transverse tubule formation, inhibits physiological hypertrophy, and represses fetal-to-adult isoform switching, thus culminating in cardiomyocyte immaturation. Mechanistically, we find that neddylation is needed for the perinatal metabolic transition from glycolytic to oxidative metabolism in cardiomyocytes. Further, we show that HIF1α is a putative neddylation target and that inhibition of neddylation accumulates HIF1α and impairs fatty acid utilization and bioenergetics in cardiomyocytes. Together, our data show neddylation is required for cardiomyocyte maturation through promoting oxidative metabolism in the developing heart.
Collapse
Affiliation(s)
- Jianqiu Zou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Wenjuan Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Yi Lu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Juan Ayala
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Kunzhe Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Hongyi Zhou
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Jinxi Wang
- Department of Medicine, University of Iowa, 200 Hawkins Drive, CBRB 2270B, Iowa City, IA 52242, USA
| | - Weiqin Chen
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Jiliang Zhou
- Department of Medicine, University of Iowa, 200 Hawkins Drive, CBRB 2270B, Iowa City, IA 52242, USA
| | - Jie Li
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
484
|
MME + fibro-adipogenic progenitors are the dominant adipogenic population during fatty infiltration in human skeletal muscle. Commun Biol 2023; 6:111. [PMID: 36707617 PMCID: PMC9883500 DOI: 10.1038/s42003-023-04504-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/18/2023] [Indexed: 01/29/2023] Open
Abstract
Fatty infiltration, the ectopic deposition of adipose tissue within skeletal muscle, is mediated via the adipogenic differentiation of fibro-adipogenic progenitors (FAPs). We used single-nuclei and single-cell RNA sequencing to characterize FAP heterogeneity in patients with fatty infiltration. We identified an MME+ FAP subpopulation which, based on ex vivo characterization as well as transplantation experiments, exhibits high adipogenic potential. MME+ FAPs are characterized by low activity of WNT, known to control adipogenic commitment, and are refractory to the inhibitory role of WNT activators. Using preclinical models for muscle damage versus fatty infiltration, we show that many MME+ FAPs undergo apoptosis during muscle regeneration and differentiate into adipocytes under pathological conditions, leading to a reduction in their abundance. Finally, we utilized the varying fat infiltration levels in human hip muscles and found less MME+ FAPs in fatty infiltrated human muscle. Altogether, we have identified the dominant adipogenic FAP subpopulation in skeletal muscle.
Collapse
|
485
|
Simulated microgravity reduces quality of ovarian follicles and oocytes by disrupting communications of follicle cells. NPJ Microgravity 2023; 9:7. [PMID: 36690655 PMCID: PMC9870914 DOI: 10.1038/s41526-023-00248-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 01/10/2023] [Indexed: 01/25/2023] Open
Abstract
Ovarian follicles are the fundamental structures that support oocyte development, and communications between oocytes and follicle somatic cells are crucial for oogenesis. However, it is unknown that whether exposure to microgravity influences cellular communications and ovarian follicle development, which might be harmful for female fertility. By 3D culturing of ovarian follicles under simulated microgravity (SMG) conditions in a rotating cell culture system, we found that SMG treatment did not affect the survival or general growth of follicles but decreased the quality of cultured follicles released oocytes. Ultrastructure detections by high-resolution imaging showed that the development of cellular communicating structures, including granulosa cell transzonal projections and oocyte microvilli, were markedly disrupted. These abnormalities caused chaotic polarity of granulosa cells (GCs) and a decrease in oocyte-secreted factors, such as Growth Differentiation Factor 9 (GDF9), which led to decreased quality of oocytes in these follicles. Therefore, the quality of oocytes was dramatically improved by the supplementations of GDF9 and NADPH-oxidase inhibitor apocynin. Together, our results suggest that exposure to simulated microgravity impairs the ultrastructure of ovarian follicles. Such impairment may affect female fertility in space environment.
Collapse
|
486
|
Furtado J, Geraldo LH, Leser FS, Poulet M, Park H, Pibouin-Fragner L, Eichmann A, Boyé K. Netrin-1 binding to Unc5B regulates Blood-Retina Barrier integrity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.21.525006. [PMID: 36711611 PMCID: PMC9882365 DOI: 10.1101/2023.01.21.525006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background The blood brain barrier (BBB) preserves neuronal function in the central nervous system (CNS) by tightly controlling metabolite exchanges with the blood. In the eye, the retina is likewise protected by the blood-retina barrier (BRB) to maintain phototransduction. We showed that the secreted guidance cue Netrin-1 regulated BBB integrity, by binding to endothelial Unc5B and regulating canonical β-catenin dependent expression of BBB gene expression. Objective Here, we investigated if Netrin-1-binding to endothelial Unc5B also controlled BRB integrity, and if this process involved Norrin/β-catenin signaling, which is the major known driver of BRB development and maintenance. Methods We analyzed Tamoxifen-inducible loss- and gain- of-function alleles of Unc5B, Ntn1 and Ctnnb1 in conjunction with tracer injections and biochemical signaling studies. Results Inducible endothelial Unc5B deletion, and inducible global Ntn1 deletion in postnatal mice reduced phosphorylation of the Norrin receptor LRP5, leading to reduced β-catenin and LEF1 expression, conversion of retina endothelial cells from a barrier-competent Claudin-5+/PLVAP- state to a Claudin-5-/PLVAP+ leaky phenotype, and extravasation of injected low molecular weight tracers. Inducible Ctnnb1 gain of function rescued vascular leak in Unc5B mutants, and Ntn1 overexpression induced BRB tightening. Unc5B expression in pericytes contributed to BRB permeability, via regulation of endothelial Unc5B. Mechanistically, Netrin-1-Unc5B signaling promoted β-catenin dependent BRB signaling by enhancing phosphorylation of the Norrin receptor LRP5 via the Discs large homologue 1 (Dlg1) intracellular scaffolding protein. Conclusions The data identify Netrin1-Unc5B as novel regulators of BRB integrity, with implications for diseases associated with BRB disruption.
Collapse
Affiliation(s)
- Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
| | - Luiz Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
| | | | - Mathilde Poulet
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, France
| | - Hyojin Park
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
| | | | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, France
| | - Kevin Boyé
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, France
| |
Collapse
|
487
|
Chen S, Shao F, Zeng J, Guo S, Wang L, Sun H, Lei JH, Lyu X, Gao S, Chen Q, Miao K, Xu X, Deng CX. Cullin-5 deficiency orchestrates the tumor microenvironment to promote mammary tumor development through CREB1-CCL2 signaling. SCIENCE ADVANCES 2023; 9:eabq1395. [PMID: 36662868 PMCID: PMC9858512 DOI: 10.1126/sciadv.abq1395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Breast cancer-associated gene 1 (Brca1) deficiency induces the onset of breast cancer formation, accompanied with extensive genetic alterations. Here, we used both the sleeping beauty transposon mutagenesis system and CRISPR-Cas9-mediated genome-wide screening in mice to identify potential genetic alterations that act synergistically with Brca1 deficiency to promote tumorignesis. Both approaches identified Cullin-5 as a tumor suppressor, whose mutation enabled Brca1-deficient cell survival and accelerated tumorigenesis by orchestrating tumor microenvironment. Cullin-5 suppresses cell growth through ubiquitylating and degrading adenosine 3',5'-monophosphate-responsive element binding protein 1 (CREB1), especially under protein damage condition. Meanwhile, Cullin-5 deficiency activated CREB1-CCL2 signaling and resulted in the accumulation of monocytes and polymorphonuclear myeloid-derived suppressor cells, reduction of T cells that benefit tumor progression in both Brca1-deficient cells and wild-type cells. Blocking CREB1 activity either through gene knockout or specific inhibitor treatment suppressed changes in the tumor microenvironment caused by Cullin-5 deficiency and blocked tumor progression.
Collapse
Affiliation(s)
- Si Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Fangyuan Shao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jianming Zeng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Sen Guo
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lijian Wang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Heng Sun
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontiers Science Center for Precision Oncogene, University of Macau, Macau SAR, China
| | - Josh Haipeng Lei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xueying Lyu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Shuai Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiang Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontiers Science Center for Precision Oncogene, University of Macau, Macau SAR, China
| | - Kai Miao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontiers Science Center for Precision Oncogene, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontiers Science Center for Precision Oncogene, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontiers Science Center for Precision Oncogene, University of Macau, Macau SAR, China
| |
Collapse
|
488
|
Zhuang M, Geng X, Han P, Che P, Liang F, Liu C, Yang L, Yu J, Zhang Z, Dong W, Ji SJ. YTHDF2 in dentate gyrus is the m 6A reader mediating m 6A modification in hippocampus-dependent learning and memory. Mol Psychiatry 2023; 28:1679-1691. [PMID: 36670199 DOI: 10.1038/s41380-023-01953-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
N6-methyladenosine (m6A) has been demonstrated to regulate learning and memory in mice. To investigate the mechanism by which m6A modification exerts its function through its reader proteins in the hippocampus, as well as to unveil the specific subregions of the hippocampus that are crucial for memory formation, we generated dentate gyrus (DG)-, CA3-, and CA1-specific Ythdf1 and Ythdf2 conditional knockout (cKO) mice, respectively. Surprisingly, we found that only the DG-specific Ythdf2 cKO mice displayed impaired memory formation, which is inconsistent with the previous report showing that YTHDF1 was involved in this process. YTHDF2 controls the stability of its target transcripts which encode proteins that regulate the elongation of mossy fibers (MF), the axons of DG granule cells. DG-specific Ythdf2 ablation caused MF overgrowth and impairment of the MF-CA3 excitatory synapse development and transmission in the stratum lucidum. Thus, this study identifies the m6A reader YTHDF2 in dentate gyrus as the only regulator that mediates m6A modification in hippocampus-dependent learning and memory.
Collapse
Affiliation(s)
- Mengru Zhuang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.,SUSTech-HKUST Joint PhD Program, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xiaoqi Geng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, China.,Department of Neurosurgery, Neurosurgical Clinical Research Center of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Peng Han
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Pengfei Che
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Fanghao Liang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Chao Liu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Lixin Yang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Jun Yu
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Zhuxia Zhang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, China. .,Department of Neurosurgery, Neurosurgical Clinical Research Center of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Sheng-Jian Ji
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
489
|
Paidi SK, Zhang Q, Yang Y, Xia CH, Ji N, Gong X. Adaptive optical two-photon fluorescence microscopy probes cellular organization of ocular lenses in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524320. [PMID: 36711806 PMCID: PMC9882239 DOI: 10.1101/2023.01.17.524320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The mammalian ocular lens is an avascular multicellular organ that grows continuously throughout life. Traditionally, its cellular organization is investigated using dissected lenses, which eliminates in vivo environmental and structural support. Here, we demonstrated that two-photon fluorescence microscopy (2PFM) can visualize lens cells in vivo. To maintain subcellular resolution at depth, we employed adaptive optics (AO) to correct aberrations due to ocular and lens tissues, which led to substantial signal and resolution improvements. Imaging lens cells up to 980 μm deep, we observed novel cellular organizations including suture-associated voids, enlarged vacuoles, and large cavities, contrary to the conventional view of a highly ordered organization. We tracked these features longitudinally over weeks and observed the incorporation of new cells during growth. Taken together, non-invasive longitudinal in vivo imaging of lens morphology using AO 2PFM will allow us to directly observe the development or alterations of lens cellular organization in living animals.
Collapse
Affiliation(s)
- Santosh Kumar Paidi
- School of Optometry, University of California, Berkeley, California 94720, USA
| | - Qinrong Zhang
- Department of Physics, University of California, Berkeley, California 94720, USA,Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Yuhan Yang
- Department of Physics, University of California, Berkeley, California 94720, USA
| | - Chun-Hong Xia
- School of Optometry, University of California, Berkeley, California 94720, USA,Vision Science Program, University of California, Berkeley, California 94720, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, California 94720, USA,Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA,Corresponding authors: Xiaohua Gong () and Na Ji ()
| | - Xiaohua Gong
- School of Optometry, University of California, Berkeley, California 94720, USA,Vision Science Program, University of California, Berkeley, California 94720, USA,Corresponding authors: Xiaohua Gong () and Na Ji ()
| |
Collapse
|
490
|
Tani H, Sadahiro T, Yamada Y, Isomi M, Yamakawa H, Fujita R, Abe Y, Akiyama T, Nakano K, Kuze Y, Seki M, Suzuki Y, Fujisawa M, Sakata-Yanagimoto M, Chiba S, Fukuda K, Ieda M. Direct Reprogramming Improves Cardiac Function and Reverses Fibrosis in Chronic Myocardial Infarction. Circulation 2023; 147:223-238. [PMID: 36503256 DOI: 10.1161/circulationaha.121.058655] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Because adult cardiomyocytes have little regenerative capacity, resident cardiac fibroblasts (CFs) synthesize extracellular matrix after myocardial infarction (MI) to form fibrosis, leading to cardiac dysfunction and heart failure. Therapies that can regenerate the myocardium and reverse fibrosis in chronic MI are lacking. The overexpression of cardiac transcription factors, including Mef2c/Gata4/Tbx5/Hand2 (MGTH), can directly reprogram CFs into induced cardiomyocytes (iCMs) and improve cardiac function under acute MI. However, the ability of in vivo cardiac reprogramming to repair chronic MI with established scars is undetermined. METHODS We generated a novel Tcf21iCre/reporter/MGTH2A transgenic mouse system in which tamoxifen treatment could induce both MGTH and reporter expression in the resident CFs for cardiac reprogramming and fibroblast lineage tracing. We first tested the efficacy of this transgenic system in vitro and in vivo for acute MI. Next, we analyzed in vivo cardiac reprogramming and fusion events under chronic MI using Tcf21iCre/Tomato/MGTH2A and Tcf21iCre/mTmG/MGTH2A mice, respectively. Microarray and single-cell RNA sequencing were performed to determine the mechanism of cardiac repair by in vivo reprogramming. RESULTS We confirmed the efficacy of transgenic in vitro and in vivo cardiac reprogramming for acute MI. In chronic MI, in vivo cardiac reprogramming converted ≈2% of resident CFs into iCMs, in which a majority of iCMs were generated by means of bona fide cardiac reprogramming rather than by fusion with cardiomyocytes. Cardiac reprogramming significantly improved myocardial contraction and reduced fibrosis in chronic MI. Microarray analyses revealed that the overexpression of MGTH activated cardiac program and concomitantly suppressed fibroblast and inflammatory signatures in chronic MI. Single-cell RNA sequencing demonstrated that resident CFs consisted of 7 subclusters, in which the profibrotic CF population increased under chronic MI. Cardiac reprogramming suppressed fibroblastic gene expression in chronic MI by means of conversion of profibrotic CFs to a quiescent antifibrotic state. MGTH overexpression induced antifibrotic effects partly by suppression of Meox1, a central regulator of fibroblast activation. CONCLUSIONS These results demonstrate that cardiac reprogramming could repair chronic MI by means of myocardial regeneration and reduction of fibrosis. These findings present opportunities for the development of new therapies for chronic MI and heart failure.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (H.T., H.Y., K.F.)
| | - Taketaro Sadahiro
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Yu Yamada
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Mari Isomi
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Hiroyuki Yamakawa
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (H.T., H.Y., K.F.)
| | - Ryo Fujita
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan.,Faculty of Medicine, and Division of Regenerative Medicine, Transborder Medical Research Center (R.F.), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Yuto Abe
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Tatsuya Akiyama
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan.,Respiratory Medicine (T.A.), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Koji Nakano
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Yuta Kuze
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Chiba, Japan (Y.K., M.S., Y.S.)
| | - Masahide Seki
- Departments of Cardiology (T.S., Y.Y., M. Isomi, R.F., Y.A., T.A., K.N., M. Ieda), University of Tsukuba, Tsukuba City, Ibaraki, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Chiba, Japan (Y.K., M.S., Y.S.)
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Chiba, Japan (Y.K., M.S., Y.S.)
| | - Manabu Fujisawa
- Hematology (M.F., M.S.-Y., S.C.), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | | | - Shigeru Chiba
- Hematology (M.F., M.S.-Y., S.C.), University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (H.T., H.Y., K.F.)
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (H.T., H.Y., K.F.)
| |
Collapse
|
491
|
Jiang Y, Wiersch J, Wu W, Qian J, Adama MPR, Wu N, Yang W, Chen C, Zhu L, Prasadan K, Gittes GK, Xiao X. Bone-marrow derived cells do not contribute to new beta-cells in the inflamed pancreas. Front Immunol 2023; 14:1084056. [PMID: 36733483 PMCID: PMC9887320 DOI: 10.3389/fimmu.2023.1084056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
The contribution of bone-marrow derived cells (BMCs) to a newly formed beta-cell population in adults is controversial. Previous studies have only used models of bone marrow transplantation from sex-mismatched donors (or other models of genetic labeling) into recipient animals that had undergone irradiation. This approach suffers from the significant shortcoming of the off-target effects of irradiation. Partial pancreatic duct ligation (PDL) is a mouse model of acute pancreatitis with a modest increase in beta-cell number. However, the possibility that recruited BMCs in the inflamed pancreas may convert into beta-cells has not been examined. Here, we used an irradiation-free model to track the fate of the BMCs from the donor mice. A ROSA-mTmG red fluorescent mouse was surgically joined to an INS1Cre knock-in mouse by parabiosis to establish a mixed circulation. PDL was then performed in the INS1Cre mice 2 weeks after parabiosis, which was one week after establishment of the stable blood chimera. The contribution of red cells from ROSA-mTmG mice to beta-cells in INS1Cre mouse was evaluated based on red fluorescence, while cell fusion was evaluated by the presence of green fluorescence in beta-cells. We did not detect any red or green insulin+ cells in the INS1Cre mice, suggesting that there was no contribution of BMCs to the newly formed beta-cells, either by direct differentiation, or by cell fusion. Thus, the contribution of BMCs to beta-cells in the inflamed pancreas should be minimal, if any.
Collapse
Affiliation(s)
- Yinan Jiang
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - John Wiersch
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Wei Wu
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Jieqi Qian
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Ultrasound in Medicine, the Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Maharana Prathap R. Adama
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nannan Wu
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Weixia Yang
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Congde Chen
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Pediatric Surgery, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingyan Zhu
- Department of Endocrinology, the First Affiliated Hospital of NanChang University, Nanchang, China
| | - Krishna Prasadan
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - George K. Gittes
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Xiangwei Xiao
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
492
|
Chen L, Dupre A, Qiu X, Pellon-Cardenas O, Walton KD, Wang J, Perekatt AO, Hu W, Spence JR, Verzi MP. TGFB1 Induces Fetal Reprogramming and Enhances Intestinal Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523825. [PMID: 36711781 PMCID: PMC9882197 DOI: 10.1101/2023.01.13.523825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The adult gut epithelium has a remarkable ability to recover from damage. To achieve cellular therapies aimed at restoring and/or replacing defective gastrointestinal tissue, it is important to understand the natural mechanisms of tissue regeneration. We employed a combination of high throughput sequencing approaches, mouse genetic models, and murine and human organoid models, and identified a role for TGFB signaling during intestinal regeneration following injury. At 2 days following irradiation (IR)-induced damage of intestinal crypts, a surge in TGFB1 expression is mediated by monocyte/macrophage cells at the location of damage. Depletion of macrophages or genetic disruption of TGFB-signaling significantly impaired the regenerative response following irradiation. Murine intestinal regeneration is also characterized by a process where a fetal transcriptional signature is induced during repair. In organoid culture, TGFB1-treatment was necessary and sufficient to induce a transcriptomic shift to the fetal-like/regenerative state. The regenerative response was enhanced by the function of mesenchymal cells, which are also primed for regeneration by TGFB1. Mechanistically, integration of ATAC-seq, scRNA-seq, and ChIP-seq suggest that a regenerative YAP-SOX9 transcriptional circuit is activated in epithelium exposed to TGFB1. Finally, pre-treatment with TGFB1 enhanced the ability of primary epithelial cultures to engraft into damaged murine colon, suggesting promise for the application of the TGFB-induced regenerative circuit in cellular therapy.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Abigail Dupre
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Xia Qiu
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Oscar Pellon-Cardenas
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Katherine D. Walton
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jianming Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Ansu O. Perekatt
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Jason R. Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Michael P. Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition & Health, Rutgers University, New Brunswick, NJ, USA
- Member of the NIEHS Center for Environmental Exposures and Disease (CEED), Rutgers EOHSI Piscataway, NJ, USA
- Lead Contact
| |
Collapse
|
493
|
Houthuijzen JM, de Bruijn R, van der Burg E, Drenth AP, Wientjens E, Filipovic T, Bullock E, Brambillasca CS, Pulver EM, Nieuwland M, de Rink I, van Diepen F, Klarenbeek S, Kerkhoven R, Brunton VG, Scheele CLGJ, Boelens MC, Jonkers J. CD26-negative and CD26-positive tissue-resident fibroblasts contribute to functionally distinct CAF subpopulations in breast cancer. Nat Commun 2023; 14:183. [PMID: 36635273 PMCID: PMC9837080 DOI: 10.1038/s41467-023-35793-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are abundantly present in the microenvironment of virtually all tumors and strongly impact tumor progression. Despite increasing insight into their function and heterogeneity, little is known regarding the origin of CAFs. Understanding the origin of CAF heterogeneity is needed to develop successful CAF-based targeted therapies. Through various transplantation studies in mice, we show that CAFs in both invasive lobular breast cancer and triple-negative breast cancer originate from mammary tissue-resident normal fibroblasts (NFs). Single-cell transcriptomics, in vivo and in vitro studies reveal the transition of CD26+ and CD26- NF populations into inflammatory CAFs (iCAFs) and myofibroblastic CAFs (myCAFs), respectively. Functional co-culture experiments show that CD26+ NFs transition into pro-tumorigenic iCAFs which recruit myeloid cells in a CXCL12-dependent manner and enhance tumor cell invasion via matrix-metalloproteinase (MMP) activity. Together, our data suggest that CD26+ and CD26- NFs transform into distinct CAF subpopulations in mouse models of breast cancer.
Collapse
Affiliation(s)
- Julia M Houthuijzen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Roebi de Bruijn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anne Paulien Drenth
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ellen Wientjens
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tamara Filipovic
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esme Bullock
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Chiara S Brambillasca
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emilia M Pulver
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marja Nieuwland
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Iris de Rink
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frank van Diepen
- Flow Cytometry Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ron Kerkhoven
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Valerie G Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Colinda L G J Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mirjam C Boelens
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
494
|
Beckmann D, Langnaese K, Gottfried A, Hradsky J, Tedford K, Tiwari N, Thomas U, Fischer KD, Korthals M. Ca 2+ Homeostasis by Plasma Membrane Ca 2+ ATPase (PMCA) 1 Is Essential for the Development of DP Thymocytes. Int J Mol Sci 2023; 24:ijms24021442. [PMID: 36674959 PMCID: PMC9865543 DOI: 10.3390/ijms24021442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
The strength of Ca2+ signaling is a hallmark of T cell activation, yet the role of Ca2+ homeostasis in developing T cells before expressing a mature T cell receptor is poorly understood. We aimed to unveil specific functions of the two plasma membrane Ca2+ ATPases expressed in T cells, PMCA1 and PMCA4. On a transcriptional and protein level we found that PMCA4 was expressed at low levels in CD4-CD8- double negative (DN) thymocytes and was even downregulated in subsequent stages while PMCA1 was present throughout development and upregulated in CD4+CD8+ double positive (DP) thymocytes. Mice with a targeted deletion of Pmca1 in DN3 thymocytes had an almost complete block of DP thymocyte development with an accumulation of DN4 thymocytes but severely reduced numbers of CD8+ immature single positive (ISP) thymocytes. The DN4 thymocytes of these mice showed strongly elevated basal cytosolic Ca2+ levels and a pre-mature CD5 expression, but in contrast to the DP thymocytes they were only mildly prone to apoptosis. Surprisingly, mice with a germline deletion of Pmca4 did not show any signs of altered progression through the developmental thymocyte stages, nor altered Ca2+ homeostasis throughout this process. PMCA1 is, therefore, non-redundant in keeping cellular Ca2+ levels low in the early thymocyte development required for the DN to DP transition.
Collapse
Affiliation(s)
- David Beckmann
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Kristina Langnaese
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Anna Gottfried
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Johannes Hradsky
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Kerry Tedford
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Nikhil Tiwari
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39120 Magdeburg, Germany
| | - Ulrich Thomas
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39120 Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| | - Mark Korthals
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
495
|
Faust TE, Feinberg PA, O’Connor C, Kawaguchi R, Chan A, Strasburger H, Masuda T, Amann L, Knobeloch KP, Prinz M, Schaefer A, Schafer DP. A comparative analysis of microglial inducible Cre lines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.09.523268. [PMID: 36711492 PMCID: PMC9881995 DOI: 10.1101/2023.01.09.523268] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cre/LoxP technology has revolutionized genetic studies and allowed for spatial and temporal control of gene expression in specific cell types. The field of microglial biology has particularly benefited from this technology as microglia have historically been difficult to transduce with virus or electroporation methods for gene delivery. Here, we interrogate four of the most widely available microglial inducible Cre lines. We demonstrate varying degrees of recombination efficiency and spontaneous recombination, depending on the Cre line and loxP distance. We also establish best practice guidelines and protocols to measure recombination efficiency in microglia, which could be extended to other cell types. There is increasing evidence that microglia are key regulators of neural circuit structure and function. Microglia are also major drivers of a broad range of neurological diseases. Thus, reliable manipulation of their function in vivo is of utmost importance. Identifying caveats and benefits of all tools and implementing the most rigorous protocols are crucial to the growth of the field of microglial biology and the development of microglia-based therapeutics.
Collapse
Affiliation(s)
- Travis E. Faust
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Philip A. Feinberg
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ciara O’Connor
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Andrew Chan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai
| | | | - Takahiro Masuda
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Lukas Amann
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Anne Schaefer
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai
- Max Planck Institute for Biology of Ageing, Cologne
| | - Dorothy P. Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
496
|
An entosis-like process induces mitotic disruption in Pals1 microcephaly pathogenesis. Nat Commun 2023; 14:82. [PMID: 36604424 PMCID: PMC9816111 DOI: 10.1038/s41467-022-35719-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023] Open
Abstract
Entosis is cell cannibalism utilized by tumor cells to engulf live neighboring cells for pro- or anti-tumorigenic purposes. It is unknown whether this extraordinary cellular event can be pathogenic in other diseases such as microcephaly, a condition characterized by a smaller than normal brain at birth. We find that mice mutant for the human microcephaly-causing gene Pals1, which exhibit diminished cortices due to massive cell death, also exhibit nuclei enveloped by plasma membranes inside of dividing cells. These cell-in-cell (CIC) structures represent a dynamic process accompanied by lengthened mitosis and cytokinesis abnormalities. As shown in tumor cells, ROCK inhibition completely abrogates CIC structures and restores the normal length of mitosis. Moreover, genetic elimination of Trp53 produces a remarkable rescue of cortical size along with substantial reductions of CIC structures and cell death. These results provide a novel pathogenic mechanism by which microcephaly is produced through entotic cell cannibalism.
Collapse
|
497
|
Tsai CH, Chuang YM, Li X, Yu YR, Tzeng SF, Teoh ST, Lindblad KE, Di Matteo M, Cheng WC, Hsueh PC, Kao KC, Imrichova H, Duan L, Gallart-Ayala H, Hsiao PW, Mazzone M, Ivanesevic J, Liu X, de Visser KE, Lujambio A, Lunt SY, Kaech SM, Ho PC. Immunoediting instructs tumor metabolic reprogramming to support immune evasion. Cell Metab 2023; 35:118-133.e7. [PMID: 36599297 PMCID: PMC10375941 DOI: 10.1016/j.cmet.2022.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/06/2022] [Accepted: 11/30/2022] [Indexed: 01/05/2023]
Abstract
Immunoediting sculpts immunogenicity and thwarts host anti-tumor responses in tumor cells during tumorigenesis; however, it remains unknown whether metabolic programming of tumor cells can be guided by immunosurveillance. Here, we report that T cell-mediated immunosurveillance in early-stage tumorigenesis instructs c-Myc upregulation and metabolic reprogramming in tumor cells. This previously unexplored tumor-immune interaction is controlled by non-canonical interferon gamma (IFNγ)-STAT3 signaling and supports tumor immune evasion. Our findings uncover that immunoediting instructs deregulated bioenergetic programs in tumor cells to empower them to disarm the T cell-mediated immunosurveillance by imposing metabolic tug-of-war between tumor and infiltrating T cells and forming the suppressive tumor microenvironment.
Collapse
Affiliation(s)
- Chin-Hsien Tsai
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City, Taiwan
| | - Yu-Ming Chuang
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Xiaoyun Li
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Yi-Ru Yu
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Sheue-Fen Tzeng
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City, Taiwan
| | - Shao Thing Teoh
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Katherine E Lindblad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Wan-Chen Cheng
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Pei-Chun Hsueh
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Kung-Chi Kao
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Hana Imrichova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Wien, Austria
| | - Likun Duan
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei City, Taiwan
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Julijana Ivanesevic
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Wien, Austria
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
| | - Karin E de Visser
- Division of Tumor Biology and Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA; Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
498
|
Kocherlakota S, Swinkels D, Van Veldhoven PP, Baes M. Mouse Models to Study Peroxisomal Functions and Disorders: Overview, Caveats, and Recommendations. Methods Mol Biol 2023; 2643:469-500. [PMID: 36952207 DOI: 10.1007/978-1-0716-3048-8_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
During the last three decades many mouse lines were created or identified that are deficient in one or more peroxisomal functions. Different methodologies were applied to obtain global, hypomorph, cell type selective, inducible, and knockin mice. Whereas some models closely mimic pathologies in patients, others strongly deviate or no human counterpart has been reported. Often, mice, apparently endowed with a stronger transcriptional adaptation, have to be challenged with dietary additions or restrictions in order to trigger phenotypic changes. Depending on the inactivated peroxisomal protein, several approaches can be taken to validate the loss-of-function. Here, an overview is given of the available mouse models and their most important characteristics.
Collapse
Affiliation(s)
- Sai Kocherlakota
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniëlle Swinkels
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Paul P Van Veldhoven
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
499
|
McDonald JA, Scott L, Van Zuylekom J, Holloway S, Blyth BJ, Sutherland KD. On Target: An Intrapulmonary Transplantation Method for Modelling Lung Tumor Development in its Native Microenvironment. Methods Mol Biol 2023; 2691:31-41. [PMID: 37355535 DOI: 10.1007/978-1-0716-3331-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2023]
Abstract
The development of in vivo lung cancer models that faithfully mimic the human disease is a crucial research tool for understanding the molecular mechanisms driving tumorigenesis. Subcutaneous transplantation assays are commonly employed, likely due to their amenability to easily monitor tumor growth and the simplistic nature of the technique to deliver tumor cells. Importantly however, subcutaneous tumors grow in a microenvironment that differs from that resident within the lung. To circumvent this limitation, here we describe the development of an intrapulmonary (iPUL) orthotopic transplantation method that enables the delivery of lung cancer cells, with precision, to the left lung lobe of recipient mice. Critically, this allows for the growth of lung cancer cells within their native microenvironment. The coupling of iPUL transplantation with position emission tomography (PET) imaging permits the serial detection of tumors in vivo and serves as a powerful tool to trace lung tumor growth and dissemination over time in mouse disease models.
Collapse
Affiliation(s)
- Jackson A McDonald
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Leanne Scott
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jessica Van Zuylekom
- Models of Cancer Translational Research Centre, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Steven Holloway
- Bioservices Department, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Benjamin J Blyth
- Models of Cancer Translational Research Centre, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.
| | - Kate D Sutherland
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
500
|
Kabir I, Zhang X, Dave JM, Chakraborty R, Qu R, Chandran RR, Ntokou A, Gallardo-Vara E, Aryal B, Rotllan N, Garcia-Milian R, Hwa J, Kluger Y, Martin KA, Fernández-Hernando C, Greif DM. The age of bone marrow dictates the clonality of smooth muscle-derived cells in atherosclerotic plaques. NATURE AGING 2023; 3:64-81. [PMID: 36743663 PMCID: PMC9894379 DOI: 10.1038/s43587-022-00342-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aging is the predominant risk factor for atherosclerosis, the leading cause of death. Rare smooth muscle cell (SMC) progenitors clonally expand giving rise to up to ~70% of atherosclerotic plaque cells; however, the effect of age on SMC clonality is not known. Our results indicate that aged bone marrow (BM)-derived cells non-cell autonomously induce SMC polyclonality and worsen atherosclerosis. Indeed, in myeloid cells from aged mice and humans, TET2 levels are reduced which epigenetically silences integrin β3 resulting in increased tumor necrosis factor [TNF]-α signaling. TNFα signals through TNF receptor 1 on SMCs to promote proliferation and induces recruitment and expansion of multiple SMC progenitors into the atherosclerotic plaque. Notably, integrin β3 overexpression in aged BM preserves dominance of the lineage of a single SMC progenitor and attenuates plaque burden. Our results demonstrate a molecular mechanism of aged macrophage-induced SMC polyclonality and atherogenesis and suggest novel therapeutic strategies.
Collapse
Affiliation(s)
- Inamul Kabir
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- To whom correspondence should be addressed: or , 203-737-2040 (phone), 203-737-6118 (FAX)
| | - Xinbo Zhang
- Department of Comparative Medicine, Yale University, New Haven, CT 06511, USA
| | - Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Raja Chakraborty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Rihao Qu
- Department of Pathology, Yale University, New Haven, CT 06511, USA
| | - Rachana R. Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Aglaia Ntokou
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Eunate Gallardo-Vara
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Binod Aryal
- Department of Comparative Medicine, Yale University, New Haven, CT 06511, USA
| | - Noemi Rotllan
- Department of Comparative Medicine, Yale University, New Haven, CT 06511, USA
| | - Rolando Garcia-Milian
- Department of Bioinformatics Support Program, Yale University, New Haven, CT 06511, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Yuval Kluger
- Department of Pathology, Yale University, New Haven, CT 06511, USA
| | - Kathleen A. Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University, New Haven, CT 06511, USA
- Department of Pathology, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- To whom correspondence should be addressed: or , 203-737-2040 (phone), 203-737-6118 (FAX)
| |
Collapse
|